CN111617078B - Pharmaceutical compositions, methods and uses thereof for the treatment and/or prevention of disease - Google Patents

Pharmaceutical compositions, methods and uses thereof for the treatment and/or prevention of disease Download PDF

Info

Publication number
CN111617078B
CN111617078B CN202010098154.4A CN202010098154A CN111617078B CN 111617078 B CN111617078 B CN 111617078B CN 202010098154 A CN202010098154 A CN 202010098154A CN 111617078 B CN111617078 B CN 111617078B
Authority
CN
China
Prior art keywords
substituted
unsubstituted
antibody
compound
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202010098154.4A
Other languages
Chinese (zh)
Other versions
CN111617078A (en
Inventor
王英
李森林
宋宏梅
游泽金
何云
蔡家强
田强
薛彤彤
王利春
王晶翼
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN111617078A publication Critical patent/CN111617078A/en
Application granted granted Critical
Publication of CN111617078B publication Critical patent/CN111617078B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Abstract

The present invention relates to pharmaceutical compositions, methods and uses thereof for the treatment and/or prevention of disease. In particular, the present invention relates to IDO (indoleamine 2, 3-dioxygenase) inhibitors containing the 1,2, 5-oxadiazole structure for use in combination with other drugs in the treatment and/or prevention of disease. In particular, the present invention relates to a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof, which is useful as an IDO inhibitor, in combination with other drugs. The combined medicament has better synergistic effect on the diseases with abnormal cell proliferation than the single medicament.

Description

Pharmaceutical compositions, methods and uses thereof for the treatment and/or prevention of disease
Technical Field
The present invention relates to the use of IDO (indoleamine 2, 3-dioxygenase) inhibitors containing the 1,2, 5-oxadiazole structure in combination with other drugs or in combination for the treatment and/or prevention of diseases. In particular, the present invention relates to a compound containing a1, 2, 5-oxadiazole structure or its isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof, which is useful as an IDO inhibitor, in combination with other therapeutic drugs.
Background
Due to unlimited growth, infiltration and metastasis of malignant tumors, three major clinical treatments (surgery, radiotherapy and chemotherapy) cannot completely remove or completely kill tumor cells, and the tumor cells can escape from the surveillance of the immune system of the body through various ways, so that tumor metastasis or recurrence frequently occurs. Tumor immunotherapy is the process of enhancing the anti-tumor immunity of the tumor microenvironment by mobilizing the immune system of the body (such as inhibiting the IDO-mediated tumor immune escape mechanism), thereby controlling and killing tumor cells. Due to the characteristics of safety, effectiveness, low adverse reaction and the like, the traditional Chinese medicine composition becomes a new therapy for treating tumors after operations, radiotherapy and chemotherapy.
IDO is currently one of the most potential small molecule drug targets for tumor immunotherapy entering the clinical research phase. It is the rate-limiting enzyme that catalyzes the catabolism of tryptophan via the kynurenine pathway. In a tumor microenvironment, multiple cells highly express IDO, which can lead to tryptophan metabolism exhaustion and kynurenine level increase, thereby blocking T cell activation, inducing oxygen free radical mediated T cell apoptosis, enhancing the immune suppression effect mediated by regulatory T cells (Treg) and promoting tumor escape from the immune monitoring of organisms.
In recent years, biological macromolecules, tumor immunochemical micromolecules, kinase inhibitor drugs and other antitumor drugs are widely applied clinically. However, due to the characteristics of diversity, mutability and the like of tumor cell signaling pathways, the drugs have the problems of drug resistance, low effective response rate, poor single-use effect and the like in application. The survival rate of the patients treated by single medicine is difficult to obviously improve. The combination of multiple drugs to improve the drug resistance of single drug therapy has become a common approach in tumor therapy.
Small molecule IDO inhibitors have been used in combination with biomacromolecule drugs and chemotherapeutic agents in a number of clinical studies. After a phase III clinical trial published by Incyte inc 2018 in 4, INCB-024360 (Epacadostat) in combination with Pembrolizumab for the treatment of advanced melanoma failed, several companies adjusted the clinical study strategy to stop the study or explore new indications. Clinical trials with combinations currently in progress include INCyte Inc. INCB-024360 (Epacadostat) (phase I/II: treatment of various cancers (metastatic and colorectal, head and neck squamous cell carcinoma, metastatic or locally advanced sarcoma, advanced malignant ductal carcinoma of the pancreas, thymus, advanced or metastatic solid tumors, etc.) in combination with the programmed death molecule 1 (PD-1) antibody pembrolizumab, metastatic non-small cell lung cancer and urothelial cancer in combination with the programmed death molecule ligand 1 (PD-L1) antibody atezolizumab, recurrent glioblastoma in combination with the PD-L1 antibody avelumab); BMS-986205 (Linrodostat) from Bristol-Myers Squibb (stage III: treatment of various cancers, such as advanced renal cell carcinoma, untreated metastatic or unresectable melanoma, muscle-invasive bladder cancer, in combination with the PD-1 antibody Nivolumab); NLG-8189 (indoximod) from NewLink Genetics corporation (stage II/III: treatment of metastatic melanoma with PD-1 antibody (Pembrolizumab or Nivolumab); stage I/II: treatment of metastatic melanoma with cytotoxic T lymphocyte antigen 4 (CTLA-4) antibody ipilimumab; treatment of metastatic pancreatic cancer with PD-L1 antibody durvalumab and therapeutics such as gemcitabine and paclitaxel; and stage II: treatment of breast cancer with docetaxel). Partial clinical test results show that the effective rate and survival rate of tumor treatment can be obviously improved by combining the IDO inhibitor and the PD-1 antibody or chemotherapeutic drugs. Therefore, the IDO inhibitor and other types of drugs (biomacromolecule drugs, chemotherapeutic drugs and the like) have good application prospects in the pharmaceutical industry.
Disclosure of Invention
The present invention provides a method for preventing and/or treating diseases of abnormal cell proliferation, which comprises combining or co-administering a prophylactically and/or therapeutically effective amount of a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof with one or more (e.g., 2 or 3) other prophylactic and/or therapeutic agents.
The invention provides a method for preparing a medicament suitable for combined use, which comprises preparing a preventive and/or therapeutic effective amount of a compound containing a1, 2, 5-oxadiazole structure or an isomer, a metabolite, a stable isotope derivative, a prodrug, a pharmaceutically acceptable salt, a hydrate, a solvate, a cocrystal or a polymorph thereof and one or more (for example, 2 or 3) other preventive and/or therapeutic medicaments into a medicament for preventing and/or treating a cell proliferation abnormal disease, wherein the medicament for preventing and/or treating the cell proliferation abnormal disease is suitable for combined use or combined use.
The present invention provides a method for preventing and/or treating diseases of abnormal cell proliferation, comprising administering to a subject a prophylactically and/or therapeutically effective amount of a compound containing the 1,2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof and one or more (e.g. 2 or 3) other prophylactic and/or therapeutic drugs.
The invention also provides the use of a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof and one or more (e.g. 2 or 3) other prophylactic and/or therapeutic drugs in the preparation of a medicament for the prevention and/or treatment of a cell proliferation disorder.
The present invention also provides a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, cocrystal or polymorph thereof together with one or more (e.g., 2 or 3) other prophylactic and/or therapeutic agents for the prevention and/or treatment of a cell proliferation abnormality disease.
The present invention also provides a pharmaceutical composition comprising a compound comprising the 1,2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof, and one or more (e.g. 2 or 3) other prophylactic and/or therapeutic agents, and a pharmaceutically acceptable carrier or excipient.
The present invention also provides a patient pack comprising means for administering a metered unit dose, the patient pack comprising a compound comprising a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof, and one or more (e.g. 2 or 3) other prophylactic and/or therapeutic drugs, optionally together with means to facilitate use of the pharmaceutical composition of the invention.
The invention also provides the use of a compound containing the 1,2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof in the manufacture of a medicament for administration with one or more (e.g. 2 or 3) other prophylactic and/or therapeutic medicaments for the treatment of a disease of abnormal cell proliferation.
The invention also provides the use of said other prophylactic and/or therapeutic agent for the manufacture of a medicament for the treatment of diseases of abnormal cell proliferation administered together with a compound containing the 1,2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be a compound having a structure represented by formula I,
Figure BDA0002385910440000031
wherein:
R 1 selected from optionally substituted C 6-10 Aryl or optionally substituted 5-6-or 8-10-membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S;
R 2 ,R 3 each independently selected from H, halogen, cyano, hydroxy, nitro, substituted or unsubstituted C 1-12 Alkyl, substituted or unsubstituted C 2-12 Alkenyl, substituted or unsubstituted C 3-12 Monocyclic alkyl, substituted or unsubstituted C 5-12 Polycycloalkyl, substituted or unsubstituted 4-10 member heterocyclic radical containing at least one heteroatom selected from N, O, S, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; or R on the same or different carbon atoms 2 And R 3 、R 2 And R 2 、R 3 And R 3 May be independently linked to optionally substituted C 3-8 A cycloalkyl group or an optionally substituted 4-to 8-membered heterocyclic group;
Figure BDA0002385910440000032
part represents a single bond or a double bond;
y is selected from N (R) 4 ) m Or C (R) 5 ) n M is an integer selected from 0 to 1, n is an integer selected from 1 to 2, R 4 And R 5 Each independently selected from hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 Polycycloalkyl, substituted or unsubstituted 4-10 member heterocyclic radical containing at least one heteroatom selected from N, O, S, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; when n =2, two R 5 May be linked to the carbon atom to which it is attached to form optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-8 membered heterocyclyl; when n =2, two R 5 May be the same or different;
z is selected from N (R) 6 ) n Or C (R) 7 ) q N is an integer selected from 1 to 2, q is an integer selected from 2 to 3, R 6 And R 7 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 Polycycloalkyl, substituted or unsubstituted 4-10 member heterocyclic radical containing at least one heteroatom selected from N, O, S, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; when n =2, two R 6 May be linked to the nitrogen atom to which it is attached to form an optionally substituted 4-8 membered heterocyclic group; when q is equal to or greater than 2, R 7 May be bonded to the carbon atom to which it is bonded to form optionally substituted C 3-8 Cycloalkyl or optionally substitutedWhen a plurality of R's are present, the 4-to 8-membered heterocyclic group of 6 Or R 7 When each R is 6 May be the same or different, each R 7 May be the same or different;
w is selected from NR 8 (R 9 ) m Or C (R) 10 ) q M is an integer selected from 0 to 1, q is an integer selected from 2 to 3, R 8 、R 9 And R 10 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; or, R 8 And R 9 And the nitrogen atom to which it is attached to form an optionally substituted 4-to 10-membered heteromonocyclic group containing at least one heteroatom selected from N, O, S or an optionally substituted 4-to 10-membered heteromonocyclic group containing at least one heteroatom selected from N, O, S; or, when q is equal to or greater than 2, R 10 May be linked to the carbon atom to which it is attached to optionally substituted C 3-8 A cycloalkyl group or an optionally substituted 4-to 8-membered heterocyclic group; when there are more than one R 10 When each R is 10 May be the same or different;
x is selected from NR 13 ,CR 14 R 15 O or S;
R 11 、R 12 are respectively selected from hydrogen, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl containing 1-4 of N, O, S, the same or different5-6 or 8-10 membered heteroaryl, substituted or unsubstituted with a heteroatom; when multiple R's are present simultaneously 11 Or a plurality of R 12 When each R is 11 Each R, which may be the same or different, is 12 May be the same or different;
R 13 、R 14 and R 15 Each independently selected from H, substituted or unsubstituted C 1-4 An alkyl group; or, R 14 And R 15 And the carbon atoms to which they are attached together form a substituted or unsubstituted C 3-12 Cycloalkyl, substituted or unsubstituted 4-10 membered heterocyclyl containing at least one heteroatom selected from N, O, S;
p is an integer selected from 1 to 6;
when a plurality of m exist at the same time, each m may be the same or different;
when a plurality of n exist at the same time, each n may be the same or different;
when a plurality of q exist at the same time, each q may be the same or different;
y, W and Z are not carbon at the same time;
Figure BDA0002385910440000041
in the step (1), the first step,
Figure BDA0002385910440000042
one and only one is a double bond;
when R is 1 Is composed of
Figure BDA0002385910440000043
R 2 、R 3 Each independently selected from hydrogen, p is 2, Y is-NH-, and W is-NH 2 or-NHCH 3 Z is C (R) 7 ) q Wherein q =2,R 7 X is not O or S when independently selected from hydrogen or nitro;
when R is 1 Is composed of
Figure BDA0002385910440000044
R 2 、R 3 Each independently selected from hydrogen, p is 2, Y is-NH-, and W is-NH 2 Z is N (R) 6 ) n Wherein n =1,R 6 X is not O or S when independently selected from hydrogen or cyano;
said "substituted" means, but is not limited to, independently being substituted by: halogen, cyano, nitro, hydroxy, amino, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, C 1-4 Alkoxy radical, C 1-4 Alkylthio radical, C 3-6 Cycloalkyl radical, R 16 SO 2 ,R 16 SO,R 16 CO,R 16 R 17 NCO,R 16 R 17 NSO 2 ,R 16 OCO, wherein R 16 、R 17 Independently selected from hydrogen and C 1-4 Alkyl when multiple R are present 16 Or a plurality of R 17 When each R is 16 Each R, which may be the same or different, is 17 May be the same or different.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be a compound having a structure represented by formula II,
Figure BDA0002385910440000051
wherein:
y is selected from NR 4 Or C (R) 5 ) 2 (ii) a Wherein R is 4 And R 5 Each independently selected from hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S; or, two R 5 To the carbon atom to which it is attached to form an optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-an 8-membered heterocyclyl; wherein two R are 5 May be the same or different;
z is selected from NR 6 Or C (R) 7 ) 2 (ii) a Wherein R is 6 And R 7 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 Polycycloalkyl, substituted or unsubstituted 4-10 member heterocyclic radical containing at least one heteroatom selected from N, O, S, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S; or, two R 7 To the carbon atom to which it is attached to form an optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-8 membered heterocyclyl; wherein two R are 7 May be the same or different;
w is selected from NR 8 R 9 Or C (R) 10 ) 3 (ii) a Wherein R is 8 、R 9 And R 10 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; or, R 8 And R 9 And the nitrogen atom to which it is attached are linked to form an optionally substituted 4-to 10-membered heteromonocyclic group containing at least one heteroatom selected from N, O, S or an optionally substituted 4-to 10-membered heteromonocyclic group containing at least one heteroatom selected from N, O, S; orTwo or three R 10 To which the carbon atom to which it is attached is linked to optionally substituted C 3-8 A cycloalkyl group or an optionally substituted 4-to 8-membered heterocyclic group; wherein each R is 10 May be the same or different;
y, W and Z are not carbon at the same time;
when R is 1 Is composed of
Figure BDA0002385910440000052
R 2 、R 3 Each independently selected from hydrogen, p is 2, Y is-NH-, and W is-NH 2 or-NHCH 3 Z is C (R) 7 ) 2 Wherein R is 7 X is not O or S when independently selected from hydrogen or nitro;
when R is 1 Is composed of
Figure BDA0002385910440000053
R 2 、R 3 Each independently selected from hydrogen, p is 2, Y is-NH-, and W is-NH 2 Z is NR 6 Wherein R is 6 X is not O or S when independently selected from hydrogen or cyano;
X,R 1 ,R 2 ,R 3 ,R 11 ,R 12 and p is as defined for formula I.
In certain embodiments, X in formula I or formula II is selected from NR 13 ,O,S,R 13 As defined in formula I or formula II.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be a compound having a structure represented by formula III,
Figure BDA0002385910440000061
wherein:
y is selected from NR 4 Or C (R) 5 ) 2 (ii) a Wherein R is 4 And R 5 Each independently selected from hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted orUnsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S; or, two R 5 To the carbon atom to which it is attached to form optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-8 membered heterocyclyl; wherein two R are 5 May be the same or different;
z is selected from NR 6 Or C (R) 7 ) 2 (ii) a Wherein R is 6 And R 7 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S; or, two R 7 To the carbon atom to which it is attached to form optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-8 membered heterocyclyl; wherein two R are 7 May be the same or different;
w is selected from NR 8 R 9 Or C (R) 10 ) 3 (ii) a Wherein R is 8 、R 9 And R 10 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl radicalSubstituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S; or, R 8 And R 9 And the nitrogen atom to which it is attached are linked to form an optionally substituted 4-to 10-membered heteromonocyclic group containing at least one heteroatom selected from N, O, S or an optionally substituted 4-to 10-membered heteromonocyclic group containing at least one heteroatom selected from N, O, S; or alternatively, two or three R 10 To which the carbon atom to which it is attached is linked to optionally substituted C 3-8 A cycloalkyl group or an optionally substituted 4-to 8-membered heterocyclic group; wherein each R is 10 May be the same or different;
y, W and Z are not carbon at the same time;
x is selected from NR 13 O or S;
when R is 1 Is composed of
Figure BDA0002385910440000062
Y is-NH-and W is-NH 2 or-NHCH 3 Z is C (R) 7 ) 2 Wherein R is 7 X is not O or S when independently selected from hydrogen or nitro;
when R is 1 Is composed of
Figure BDA0002385910440000063
Y is-NH-and W is-NH 2 Z is NR 6 Wherein R is 6 X is not O or S when independently selected from hydrogen or cyano;
R 1 ,R 11 ,R 12 ,R 13 as defined in any one of formula I or formula II.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be a compound having a structure represented by formula IV,
Figure BDA0002385910440000071
wherein:
R 4 selected from hydrogen, cyano, nitro, R 11 SO 2 ,R 12 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 Polycycloalkyl, substituted or unsubstituted 4-10 member heterocyclic radical containing at least one heteroatom selected from N, O, S, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S;
z is selected from NR 6 Or C (R) 7 ) 2 (ii) a Wherein R is 6 Or R 7 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S; or, two R 7 To the carbon atom to which it is attached to form an optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-8 membered heterocyclyl; wherein two R are 7 May be the same or different;
w is selected from NR 8 R 9 Or C (R) 10 ) 3 (ii) a Wherein R is 8 、R 9 Or R 10 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl radicals, i.e. takingSubstituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; or, R 8 And R 9 And the nitrogen atom to which it is attached to form an optionally substituted 4-10 membered heteromonocyclic group containing at least one heteroatom selected from N, O, S or an optionally substituted 4-10 membered heteromonocyclic group containing at least one heteroatom selected from N, O, S; or alternatively, two or three R 10 To which the carbon atom to which it is attached is linked to optionally substituted C 3-8 A cycloalkyl group or an optionally substituted 4-to 8-membered heterocyclic group; wherein each R is 10 May be the same or different;
x is selected from NR 13 O or S;
when R is 1 Is composed of
Figure BDA0002385910440000072
Y is-NH-and W is-NH 2 or-NHCH 3 Z is C (R) 7 ) 2 Wherein R is 7 X is not O or S when independently selected from hydrogen or nitro;
when R is 1 Is composed of
Figure BDA0002385910440000073
Y is-NH-and W is-NH 2 Z is NR 6 Wherein R is 6 X is not O or S when independently selected from hydrogen or cyano;
R 1 ,R 11 ,R 12 ,R 13 as defined in any one of formulas I-III.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be a compound having a structure represented by formula IV-1,
Figure BDA0002385910440000081
wherein:
R 6 selected from hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S;
when R is 1 Is composed of
Figure BDA0002385910440000082
Y is-NH-and W is-NH 2 ,R 6 X is not O or S when independently selected from hydrogen or cyano;
X,W,R 1 ,R 4 ,R 8 ,R 9 ,R 10 ,R 11 ,R 12 ,R 13 as defined in any one of formulas I-IV.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be a compound having a structure represented by formula IV-2,
Figure BDA0002385910440000083
wherein the content of the first and second substances,
R 7 selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S;
when R is 1 Is composed of
Figure BDA0002385910440000084
Y is-NH-and W is-NH 2 or-NHCH 3 ,R 7 X is not O or S when independently selected from hydrogen or nitro;
X,W,R 1 ,R 4 ,R 8 ,R 9 ,R 10 ,R 11 ,R 12 ,R 13 as defined in any one of formulas I-IV.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be a compound having a structure represented by formula V-1,
Figure BDA0002385910440000085
wherein:
z is selected from N (R) 6 ) 2 Or C (R) 7 ) 3 (ii) a Wherein R is 6 And R 7 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; or, two R 6 To the nitrogen atom to which it is attached to form an optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-8 membered heterocyclyl; or alternatively, two or three R 7 To which the carbon atom to which it is attached is linked to optionally substituted C 3-8 Cycloalkyl radicals or radicals optionally takenSubstituted 4-8 membered heterocyclyl; wherein each R is 6 May be the same or different; each R is 7 May be the same or different;
w is selected from NR 8 R 9 Or C (R) 10 ) 3 (ii) a Wherein R is 8 、R 9 And R 10 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 Polycycloalkyl, substituted or unsubstituted 4-10 member heterocyclic radical containing at least one heteroatom selected from N, O, S, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S; or, R 8 And R 9 And the nitrogen atom to which it is attached are linked to form an optionally substituted 4-to 10-membered heteromonocyclic group containing at least one heteroatom selected from N, O, S or an optionally substituted 4-to 10-membered heteromonocyclic group containing at least one heteroatom selected from N, O, S; or alternatively, two or three R 10 To which the carbon atoms to which they are attached are linked to form optionally substituted C 3-8 A cycloalkyl group or an optionally substituted 4-to 8-membered heterocyclic group; wherein each R is 10 May be the same or different;
x is selected from NR 13 O or S;
R 1 ,R 11 ,R 12 ,R 13 as defined in formula I.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be a compound having a structure represented by formula V-2,
Figure BDA0002385910440000091
wherein:
R 5 selected from hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 Polycycloalkyl, substituted or unsubstituted 4-10 member heterocyclic radical containing at least one heteroatom selected from N, O, S, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1-4 identical or different heteroatoms selected from N, O, S;
z is selected from N (R) 6 ) 2 Or C (R) 7 ) 3 (ii) a Wherein R is 6 And R 7 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; or, two R 6 To the nitrogen atom to which it is attached to form an optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-8 membered heterocyclyl; or alternatively, two or three R 7 To which the carbon atom to which it is attached is linked to optionally substituted C 3-8 Cycloalkyl or optionally substituted 4-8 membered heterocyclyl; wherein each R is 6 May be the same or different; each R is 7 May be the same or different;
w is selected from NR 8 R 9 Or C (R) 10 ) 3 (ii) a Wherein R is 8 、R 9 Or R 10 May be the same or different and are each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Monocyclic alkyl, substituted or unsubstituted C 5-10 A polycycloalkyl group, a substituted or unsubstituted 4-to 10-membered heterocyclic group containing at least one heteroatom selected from N, O and S, and a substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; or, R 8 And R 9 And the nitrogen atom to which it is attached to form an optionally substituted 4-10 membered heteromonocyclic group containing at least one heteroatom selected from N, O, S or an optionally substituted 4-10 membered heteromonocyclic group containing at least one heteroatom selected from N, O, S; or alternatively, two or three R 10 To which the carbon atoms to which they are attached are linked to form optionally substituted C 3-8 A cycloalkyl group or an optionally substituted 4-to 8-membered heterocyclic group; each R is 10 May be the same or different;
x is selected from NR 13 O or S;
R 1 ,R 11 ,R 12 ,R 13 as defined in formula I.
In certain embodiments, in the compound having a structure as shown in formula I, II, III, IV-1, IV-2, V-1, or V-2, R 1 Selected from optionally substituted C 6-10 An aryl group; preferably, R 1 Selected from optionally substituted phenyl; preferably, R 1 Selected from phenyl substituted by one or more halogens; preferably, R 1 Is selected from
Figure BDA0002385910440000101
Particularly preferably, R 1 Is composed of
Figure BDA0002385910440000102
In certain embodiments, in the compound having a structure as shown in formula I, II, III, IV-1 or IV-2, R 4 Selected from hydrogen, cyano, nitro, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 An alkenyl group; preferably, R 4 Is selected from the group consisting of hydrogen,substituted or unsubstituted C 1-4 An alkyl group; further preferably, R 4 Selected from hydrogen and methyl; particularly preferably, R 4 Is hydrogen.
In certain embodiments, the compound having a structure as shown in formula I, II, III, or IV-2, R 5 Is hydrogen.
In certain embodiments, in the compound having a structure as shown in formula I, II, III, IV-1, IV-2, V-1, or V-2, R 6 Or R 7 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Cycloalkyl, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; preferably, R 6 Or R 7 Each independently selected from: hydrogen, cyano, nitro, R 11 SO 2 ,R 11 CO,R 11 R 12 NSO 2 ,R 11 R 12 NCO,R 11 OCO, substituted or unsubstituted C 1-4 An alkyl group; preferably, R 6 Or R 7 Each independently selected from: hydrogen, cyano, nitro, methylsulfonyl, NH 2 - (C = O) -, sulfonamido, ethanesulfonyl, cyclopropylsulfonyl, isopropylsulfonyl, cyclohexylsulfonyl, phenylsulfonyl, -CH 2 CF 3 ,-CH 2 CN; particularly preferably, R 6 Or R 7 Each independently selected from: hydrogen, cyano, nitro, methylsulfonyl, NH 2 - (C = O) -, sulfonamido, ethanesulfonyl, cyclopropylsulfonyl, isopropylsulfonyl, benzenesulfonyl, -CH 2 CF 3
In certain embodiments, in the compound having a structure as shown in formula I, II or III, Y is selected from NR 4 Or C (R) 5 ) 2 (ii) a Preferably, Y is selected from NH or CH 2 (ii) a More preferably, Y is selected from NH.
In certain embodiments, in the compound having a structure as shown in formula I, II, III, IV-1, IV-2, V-1, or V-2, W is selected from NR 8 R 9 Or C (R) 10 ) 3 (ii) a Wherein R is 8 、R 9 Or R 10 May be the same or different and are each independently selected from: hydrogen, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 2-8 Alkenyl, substituted or unsubstituted C 3-8 Cycloalkyl, substituted or unsubstituted C 6-10 Aryl, substituted or unsubstituted 5-6 or 8-10 membered heteroaryl containing 1 to 4 identical or different heteroatoms selected from N, O, S; or, R 8 And R 9 And the nitrogen atom to which it is attached to form an optionally substituted 4-10 membered heteromonocyclic group containing at least one heteroatom selected from N, O, S or an optionally substituted 4-10 membered heteromonocyclic group containing at least one heteroatom selected from N, O, S; or, R 10 To which the carbon atom to which it is attached is linked to optionally substituted C 3-8 A cycloalkyl group or an optionally substituted 4-to 8-membered heterocyclic group; preferably, W is NR 8 R 9 (ii) a Wherein R is 8 Or R 9 Each independently selected from: hydrogen, substituted or unsubstituted C 1-8 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted C 6-10 An aryl group; or, R 8 And R 9 And the nitrogen atom to which it is attached to form an optionally substituted 5-6 membered heteromonocyclic group containing at least one heteroatom selected from N, O, S or an optionally substituted 4-10 membered heteromonocyclic group containing at least one heteroatom selected from N, O, S; more preferably, W is NR 8 R 9 (ii) a Wherein R is 8 Or R 9 Each independently selected from: hydrogen, substituted or unsubstituted C 1-4 Alkyl, substituted or unsubstituted C 3-6 Cycloalkyl, substituted or unsubstituted phenyl; or, R 8 And R 9 And the nitrogen atom to which it is attached to form an optionally substituted morpholinyl, piperidinyl, piperazinyl, tetrahydropyrrolyl or an optionally substituted 4-to 10-membered spiroheterocyclyl containing at least one heteroatom selected from N, O, S; particularly preferably, W is selected from amino, tert-butylamino, phenylamino, benzylamino, isopropylamino, dimethylaminoA morpholinyl group, a piperidinyl group, a tetrahydropyrrolyl group, a methylamino group, a cyclohexylamino group, an ethylamino group, a cyclopropylamino group,
Figure BDA0002385910440000111
Figure BDA0002385910440000112
In certain embodiments, in the compound having a structure as shown in formula I, II, III, IV-1, IV-2, V-1, or V-2, R 13 Selected from H, substituted or unsubstituted C 1-4 An alkyl group; preferably, R 13 Is H.
In certain embodiments, in the compound having a structure as shown in formula I, II, III, IV-1, IV-2, V-1, or V-2, Z is selected from NR 6 Or C (R) 7 ) 2 ,R 6 And R 7 Each independently selected from: cyano, nitro, R 11 SO 2 ,R 11 R 12 NSO 2 ,R 11 R 12 NCO, trifluoroethyl; r 11 、R 12 Independently selected from hydrogen, methyl, ethyl, isopropyl, cyclopropyl, phenyl; preferably, Z is CHR 7 ,R 7 Is nitro.
In certain embodiments, in the compounds having a structure according to formula I, II, III, IV-1, IV-2, V-1, or V-2, the "substitution" refers to independently substitution with: halogen, cyano, nitro, hydroxy, amino, C 1-4 Alkyl radical, C 2-4 Alkenyl radical, C 2-4 Alkynyl, C 1-4 Alkoxy radical, C 1-4 Alkylthio radical, C 3-6 Cycloalkyl radical, R 16 SO 2 ,R 16 SO,R 16 CO,R 16 R 17 NCO,R 16 R 17 NSO 2 ,R 16 OCO, wherein R 16 、R 17 Independently selected from hydrogen and C 1-4 Alkyl when multiple R are present 16 Or a plurality of R 17 When each R is 16 Each R, which may be the same or different, is 17 May be the same or different; preferably, said "substitution" means aloneIs substituted on site with the following groups: fluorine, chlorine, bromine, hydroxyl, methyl or methoxy.
In certain embodiments, in the compound having a structure as shown in formula I, II, III, IV-1, IV-2, V-1, or V-2, X is selected from NH, O, or S.
In the present invention, the compound having a1, 2, 5-oxadiazole structure may be selected from:
Figure BDA0002385910440000113
Figure BDA0002385910440000121
Figure BDA0002385910440000131
Figure BDA0002385910440000141
Figure BDA0002385910440000151
Figure BDA0002385910440000161
Figure BDA0002385910440000171
Figure BDA0002385910440000181
Figure BDA0002385910440000191
Figure BDA0002385910440000201
Figure BDA0002385910440000211
preferably, the compound containing the 1,2,5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal, or polymorph thereof is as described in PCT/CN2018/094523 (which is incorporated herein in its entirety).
More preferably, the compound containing a1, 2, 5-oxadiazole structure is selected from the compounds of the structures shown below:
Figure BDA0002385910440000212
particularly preferably, the compound containing a1, 2, 5-oxadiazole structure is selected from the compounds of the structures shown below:
Figure BDA0002385910440000213
in the present invention, the other prophylactic and/or therapeutic agents include, but are not limited to: an antitumor drug.
In the present invention, the antitumor drugs include, but are not limited to: antibody antineoplastic drugs, cytotoxic antineoplastic drugs, drugs affecting endocrine balance, biological response regulators, kinase small molecule inhibitors and other auxiliary preventive and/or therapeutic drugs.
In the invention, the antibody antineoplastic drugs include, but are not limited to, trastuzumab, rituximab, cetuximab, bevacizumab, nimotuzumab, iodine (131I) metuximab, PD-1 antibody and PD-L1 antibody.
Preferably, the PD-1 antibody is a monoclonal antibody against the PD-1 target. In certain embodiments, the PD-1 antibody is a murine PD-1 antibody or a human PD-1 antibody, e.g., selected from murine PD-1 antibody (Bioxcell, BP 0146), pembrolizumab (Keytruda), nivolumab (Opdivo), and the like.
Preferably, the PD-L1 antibody is a monoclonal antibody against a PD-L1 target and is selected from the group consisting of Atzhugumab (Teentriq), devolumab (Imfinizi), avelumab (Bavencio) and the like.
In the present invention, the cytotoxic antitumor agents include, but are not limited to: mechlorethamine, cyclophosphamide, glycerole, cisplatin, oxaliplatin, carboplatin, iproplatin, nedaplatin, leplatin, doxorubicin, pirarubicin, mitomycin, bleomycin, actinomycin-D, doxorubicin, methotrexate, fluorouracil, hydroxyurea, cytarabine, gemcitabine, azacytidine, fludarabine, nelarabine, forodesine, cladribine, clofarabine, capecitabine, mercaptopurine, vincristine, paclitaxel, homoharringtonine, asparaginase, irinotecan, etoposide, retinoic acid, bortezomib.
In the present invention, the endocrine balance affecting drugs include, but are not limited to: tamoxifen, toremifene, flutamide, letrozole, exemestane, megestrol, goserelin, triptorelin, and the like.
In the present invention, the biological response modifier drugs include, but are not limited to: interleukin-2, thymosin, interferon, and the like.
In the invention, the kinase small molecule inhibitor antitumor drugs include but are not limited to: reversible non-receptor tyrosine kinase (NRTK) inhibitor antineoplastic drugs, reversible Receptor Tyrosine Kinase (RTK) inhibitor antineoplastic drugs, irreversible protein kinase inhibitor antineoplastic drugs, serine/threonine kinase inhibitor antineoplastic drugs and lipid kinase inhibitor antineoplastic drugs.
In a preferred embodiment, the reversible non-receptor tyrosine kinase (NRTK) inhibitor class of antineoplastic drugs include, but are not limited to: sunitinib malate, dasatinib, nilotinib hydrochloride, bosutinib, ponatinib hydrochloride, lucagolide phosphate, and tofacitinib citrate.
In a preferred embodiment, the reversible receptor-type tyrosine kinase (RTK) inhibitor antineoplastic drugs include, but are not limited to: gefitinib, nilotinib, lapatinib, vandetanib, afatinib, oxitinib, sorafenib, pezopanib, axitinib, regorafenib, nidanib, lenvatinib, crizotinib, ceritinib, cabozitinib, and alitanib.
In a preferred embodiment, the irreversible protein kinase inhibitor antineoplastic agents include, but are not limited to: ibrutinib.
In a preferred embodiment, the serine/threonine kinase inhibitor antineoplastic drugs include, but are not limited to: vemurafenib, dabrafenib mesylate, trametinib dimethyl sulfoxide, kemetinib and palbociclib.
In a preferred embodiment, the lipid kinase inhibitor antineoplastic drugs include, but are not limited to: idellarib.
In preferred embodiments, the additional adjunctive prophylactic and/or therapeutic agents include, but are not limited to: recombinant human granulocyte colony stimulating factor, erythropoietin, interleukin D11, indomethacin, tramadol, morphine, domperidone, granisetron, tropisetron, pamidronate disodium, zoledronic acid and sorafenib.
In a preferred embodiment, the cell proliferation disorder is selected from the group consisting of disorders associated with IDO activity or IDO-mediated immunosuppression.
In a preferred embodiment, said cell proliferation disorder is selected from the group consisting of IDO overexpressing tumors. In a preferred embodiment, the tumor is resistant or insensitive to immunotherapy.
In the present invention, the cell proliferative disorder is selected from cancers occurring in the esophagus, stomach, intestine, rectum, oral cavity, pharynx, larynx, lung, colon, breast, uterus, endometrium, ovary, prostate, testis, bladder, kidney, liver, pancreas, bone, connective tissue, skin, eye, brain or central nervous system, and thyroid cancer, leukemia, hodgkin's disease, lymphoma and myeloma.
In a preferred embodiment, the tumor is selected from lung cancer (e.g., non-small cell lung cancer), gastric cancer, intestinal cancer, colon cancer, breast cancer, cervical cancer, rectal cancer, pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, liver cancer, fallopian tube cancer, ovarian tumor, peritoneal tumor, melanoma, glioma, hepatocellular carcinoma, papillary renal tumor, head and neck tumor, leukemia, lymphoma, myeloma, glioblastoma, pancreatic ductal carcinoma, thymus carcinoma, or metastatic solid tumor. Preferably, the tumor is a tumor expressing PD-L1, more preferably breast cancer, lung cancer (e.g., non-small cell lung cancer), gastric cancer, intestinal cancer, colon cancer, renal cancer, melanoma.
In particular, the present invention relates to a method for the prevention and/or treatment of diseases of abnormal cell proliferation comprising administering a prophylactically and/or therapeutically effective amount of a compound containing the 1,2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof in combination or association with one or more (e.g. 2 or 3) other prophylactic and/or therapeutic agents; the compound containing a1, 2, 5-oxadiazole structure is selected from the group consisting of:
Figure BDA0002385910440000231
Figure BDA0002385910440000241
preferably, the other preventive and/or therapeutic drugs are antibody-based antitumor drugs; preferably, the antibody antitumor drug is selected from a PD-1 antibody or a PD-L1 antibody; preferably, the antibody antineoplastic drug is a PD-1 antibody.
In particular, the present invention relates to a method for preparing a medicament suitable for combined use, comprising combining a prophylactically and/or therapeutically effective amount of a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof with one or more (e.g., 2 or 3) other prophylactic and/or therapeutic agents to prepare a medicament for preventing and/or treating a cell proliferation abnormality disease, which is suitable for combined use or combined use; the compound containing a1, 2, 5-oxadiazole structure is selected from:
Figure BDA0002385910440000242
preferably, the other preventive and/or therapeutic drugs are antibody antitumor drugs; preferably, the antibody antineoplastic drugs are selected from PD-1 antibodies or PD-L1 antibodies; preferably, the antibody antineoplastic drug is a PD-1 antibody.
In particular, the present invention relates to a method for preventing and/or treating diseases with abnormal cell proliferation, comprising administering to a subject a prophylactically and/or therapeutically effective amount of a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof and one or more (e.g. 2 or 3) other prophylactic and/or therapeutic drugs; the compound containing a1, 2, 5-oxadiazole structure is selected from the group consisting of:
Figure BDA0002385910440000251
preferably, the other preventive and/or therapeutic drugs are antibody antitumor drugs; preferably, the antibody antineoplastic drugs are selected from PD-1 antibodies or PD-L1 antibodies; preferably, the antibody antitumor drug is a PD-1 antibody.
In particular, the present invention relates to the use of a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof, together with one or more (e.g. 2 or 3) other prophylactic and/or therapeutic agents in the manufacture of a medicament for the prevention and/or treatment of diseases associated with abnormal cell proliferation; the compound containing a1, 2, 5-oxadiazole structure is selected from the group consisting of:
Figure BDA0002385910440000252
Figure BDA0002385910440000261
preferably, the other preventive and/or therapeutic drugs are antibody-based antitumor drugs; preferably, the antibody antitumor drug is selected from a PD-1 antibody or a PD-L1 antibody; preferably, the antibody antitumor drug is a PD-1 antibody.
In particular, the present invention relates to a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof, together with one or more (e.g. 2 or 3) other prophylactic and/or therapeutic agents for the prevention and/or treatment of diseases of abnormal cell proliferation; the compound containing a1, 2, 5-oxadiazole structure is selected from:
Figure BDA0002385910440000262
preferably, the other preventive and/or therapeutic drugs are antibody-based antitumor drugs; preferably, the antibody antitumor drug is selected from a PD-1 antibody or a PD-L1 antibody; preferably, the antibody antitumor drug is a PD-1 antibody.
In a preferred embodiment, a compound comprising the 1,2, 5-oxadiazole structure of the invention, such as compound 50, compound 50-a, compound 50-b or compound 35, or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph of said compound is selected for use in combination or co-administration with a PD-1 antibody or a PD-L1 antibody for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, renal cancer, non-small cell lung cancer, pancreatic ductal carcinoma, thymus carcinoma or metastatic solid tumors.
In certain embodiments, compound 50-a of the present invention, or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal, or polymorph thereof, is selected for use in combination or co-administration with a PD-1 antibody for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, renal cancer, non-small cell lung cancer, ductal pancreatic cancer, thymus cancer, or metastatic solid tumors; preferably, for the prevention and/or treatment of colon cancer.
In certain embodiments, compound 35 of the present invention, or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal, or polymorph thereof, is selected for use in combination or co-administration with a PD-1 antibody for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, renal cancer, non-small cell lung cancer, ductal pancreatic cancer, thymus cancer, or metastatic solid tumors; preferably, for the prevention and/or treatment of colon cancer.
In certain embodiments, compound 50-a of the present invention, or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal, or polymorph thereof, is selected for use in combination or co-administration with a PD-L1 antibody for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, renal cancer, non-small cell lung cancer, ductal pancreatic cancer, thymus cancer, or metastatic solid tumors; preferably, for the prevention and/or treatment of colon cancer.
In certain embodiments, compound 35 of the present invention, or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal, or polymorph thereof, is selected for use in combination or co-administration with a PD-L1 antibody for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, renal cancer, non-small cell lung cancer, ductal pancreatic cancer, thymus cancer, or metastatic solid tumors; preferably, for the prevention and/or treatment of colon cancer.
The invention also relates to the use of compound 50-a or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof together with a PD-1 antibody for the manufacture of a medicament for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, renal cancer, non-small cell lung cancer, ductal carcinoma of the pancreas, thymus carcinoma or metastatic solid tumors; preferably, in the manufacture of a medicament for the prevention and/or treatment of colon cancer.
The invention also relates to the use of compound 35 or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof together with a PD-1 antibody for the manufacture of a medicament for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, kidney cancer, non-small cell lung cancer, ductal carcinoma of the pancreas, thymus carcinoma or metastatic solid tumors; preferably, for the preparation of a medicament for the prevention and/or treatment of colon cancer.
The invention also relates to the use of compound 50-a or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof with an antibody PD-L1 for the manufacture of a medicament for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, renal cancer, non-small cell lung cancer, ductal carcinoma of the pancreas, thymus carcinoma or metastatic solid tumors; preferably, in the manufacture of a medicament for the prevention and/or treatment of colon cancer.
The invention also relates to the use of compound 35 or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof and a PD-L1 antibody for the manufacture of a medicament for the prevention and/or treatment of colon cancer, melanoma, rectal cancer, glioblastoma, renal cancer, non-small cell lung cancer, pancreatic ductal carcinoma, thymus carcinoma or metastatic solid tumors; preferably, for the preparation of a medicament for the prevention and/or treatment of colon cancer.
In the present invention, the pharmaceutical composition of the invention may be a pharmaceutical product (combination) in one or more dosage units to obtain a suitable combination. The pharmaceutical product may be administered to a patient or subject, such as a mammal (preferably a human), in need thereof. The compound containing the 1,2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph thereof and the one or more (e.g. 2 or 3) other prophylactic and/or therapeutic drugs in the pharmaceutical product may be present in the same dosage unit or in different dosage units, which may be used simultaneously, sequentially or alternately.
Definition of terms
Unless defined otherwise below, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art. Reference to the techniques used herein is intended to refer to techniques commonly understood in the art, including those variations of or alternatives to those techniques that would be apparent to one of ordinary skill in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
The terms "comprising," "including," "having," "containing," or "involving," and other variants thereof herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps.
In the present invention, the term "combination" or "administration of a combination" encompasses the simultaneous, sequential, and alternating use of two or more drugs, which comprises formulating the two or more drugs in one or more dosage units to obtain a drug product suitable for administration of the combination, and administering the drug product to a mammal in need of such combination. The "sequentially" use includes: administering a compound comprising the 1,2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal, or polymorph thereof, followed by administration of another prophylactic and/or therapeutic agent; further comprising: the other prophylactic and/or therapeutic drugs are administered first, followed by the compound containing the 1,2, 5-oxadiazole structure or its isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph.
The drug or pharmaceutical composition of the present invention may be in the form of a solid preparation, a semisolid preparation, a liquid preparation, a gaseous preparation, or the like. For example, the solid preparation is tablets, capsules, powder, granules or suppositories, and the like, and the liquid preparation is solutions, suspensions or injections. The composition can also be in the form of liposome, microsphere, etc. In particular, the pharmaceutical composition is in a form of a formulation suitable for oral administration, or in a form of a formulation for injection administration.
The term "pharmaceutically acceptable carrier" as used herein refers to an inactive ingredient in a pharmaceutical composition or pharmaceutical formulation that does not cause significant irritation to an organism and does not substantially affect the biological activity of the administered active ingredient, and includes, for example, diluents, adjuvants, excipients or suitable pharmaceutically acceptable media. Examples of suitable pharmaceutically acceptable carriers are described in Remington's Pharmaceutical Sciences (1990).
The term "metabolite form" as used herein refers to a compound produced in vivo after administration to an individual in need thereof.
The term "pharmaceutically acceptable salts" as used herein includes both acid and base addition salts thereof. Such as hexafluorophosphate, meglumine salt, and the like. For a review of suitable Salts, see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: properties, selection, and Use" (Wiley-VCH, 2002).
The medicaments according to the invention, such as the compounds containing the 1,2, 5-oxadiazole structure and/or the other prophylactic and/or therapeutic medicaments, can be present in the form of hydrates or solvates, wherein the medicament or active ingredient according to the invention comprises a solvent, such as, in particular, water, ethanol, as a structural element of the crystal lattice of the compound.
The present invention encompasses all possible crystalline forms or polymorphs of the medicament of the present invention (including the active ingredient contained therein), which may be a single polymorph or a mixture of more than one polymorph in any ratio.
In the present invention, the compounds containing the 1,2, 5-oxadiazole structure are intended to exist in the form of stereoisomers (which include cis and trans isomers, optical isomers (e.g., R and S enantiomers), diastereomers, geometric isomers, rotamers, conformational isomers, atropisomers, and mixtures thereof). The compounds may exhibit more than one type of isomerization and consist of mixtures thereof (e.g., racemic mixtures and diastereomeric pairs).
The term "excipient" as used herein refers to a substance used in the preparation of pharmaceutical compositions that is generally safe and neither biologically nor otherwise undesirable, and includes a variety of excipients suitable for veterinary use as well as human pharmaceutical use.
In the pharmaceutical compositions of the present invention, pharmaceutically acceptable carriers that can be employed include, but are not limited to, sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
Water is an exemplary carrier when the pharmaceutical composition is administered intravenously. Physiological saline and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like. The composition may also optionally contain minor amounts of wetting agents, emulsifying agents or pH buffering agents. Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like.
The term "formulation" or "dosage form" as used herein is intended to include solid, semi-solid, liquid and gaseous formulations of the compounds of the present invention. Such formulations or dosage forms include, but are not limited to, tablets, capsules, lozenges, hard candies, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions, injectable solutions, elixirs, syrups. One skilled in the art will appreciate that the compounds of the present invention may be formulated into different formulations depending on the desired dosage and pharmacokinetic parameters.
The unit dose range of the pharmaceutically active ingredient of the present invention may be 0.1-1000mg, preferably 1-800mg, more preferably 10-600mg, especially preferably 50-450mg, and most preferably 100-300mg.
In the present invention, the unit dose of the pharmaceutically active ingredient (e.g., a compound containing a1, 2, 5-oxadiazole structure or an isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, cocrystal or polymorph thereof, or other prophylactic and/or therapeutic drug) is not particularly limited, and is, for example, administered in an amount of about 1mg, about 2mg, about 3mg, about 4mg, about 5mg, about 6mg, about 7mg, about 8mg, about 9mg, about 10mg, about 25mg, about 50mg, about 75mg, about 100mg, about 150mg, about 200mg, about 250mg, about 300mg, about 350mg, about 400mg, about 500mg, or about 600 mg. The formulation or dosage form of the present invention may contain a single or multiple unit doses of the pharmaceutically active ingredient of the present invention as described above.
In the present invention, the active ingredient PD-1 antibody or PD-L1 antibody can be administered parenterally. In the present invention, the unit dose of the PD-1 antibody or the PD-L1 antibody is not particularly limited, and is administered, for example, in the following amounts: about 0.5mg/Kg (about 0.5mg of anti-PD-L1 monoclonal antibody per kilogram of the subject mass), about 1mg/Kg, about 2mg/Kg, about 3mg/Kg, about 4mg/Kg, about 5mg/Kg, about 6mg/Kg, about 7mg/Kg, about 8mg/Kg, about 9mg/Kg, about 10mg/Kg, about 11mg/Kg, about 12mg/Kg, about 13mg/Kg, about 14mg/Kg, about 15mg/Kg, about 16mg/Kg, about 17mg/Kg, about 18mg/Kg, about 19mg/Kg, or about 20mg/Kg. In a particular embodiment, the PD-1 antibody is administered intravenously in the dosage amounts described above.
The medicament of the present invention is preferably administered orally. Depending on the circumstances, other routes of administration may be used or even preferred, such as intravenous, intra-arterial, subcutaneous, intraperitoneal, intramuscular or transdermal administration; or by buccal, nasal, transmucosal, topical, in the form of ophthalmic preparations or by inhalation, and the like. Transdermal administration may be highly desirable for patients who are forgetful or who are poorly tolerated with oral medications. In particular cases, the compounds of the invention may also be administered by the transdermal, intramuscular, intranasal or intrarectal route. The route of administration may vary in any manner, depending on the physical characteristics of the drug, the convenience of the patient and caregiver, and other relevant circumstances (Remington's Pharmaceutical Sciences, 18 th edition, mack Publishing co. (1990)).
The active ingredient of the present invention or a product (e.g., a pharmaceutical composition, a pharmaceutical preparation or a dosage form) comprising the same may be administered in a dose range of 0.1 to 1000mg/kg body weight/day, preferably in a dose range of 0.1 to 800mg/kg body weight/day, preferably in a dose range of 1 to 600mg/kg body weight/day, preferably in a dose range of 10 to 400mg/kg body weight/day, particularly preferably in a dose range of 50 to 300mg/kg body weight/day, and most preferably in a dose range of 100 to 250mg/kg body weight/day. The precise dose required to prevent and/or treat a patient is determined by a physician based on the stage and severity of the disease and the specific needs and response of the individual patient.
As used herein, unless otherwise specified, the term "treating" means reversing, alleviating, inhibiting the progression of, or preventing such a disorder or condition, or one or more symptoms of such a disorder or condition, to which such term applies. The term "preventing" means preventing additional symptoms, preventing a potential metabolic predisposition for a symptom, inhibiting the disease or disorder, e.g., arresting the occurrence of the disease or disorder.
In the present invention, the term "PD-1" may also be referred to as "Programmed death molecule 1", "protein PD-1", "hPD-I", and may be used interchangeably.
In the present invention, the term "PD-L1" may also be referred to as "Programmed death-ligand 1", "Programmed cell death-ligand 1", "protein PD-L1", "PDL1", "PDCDL1", "hPD-L1", "hPD-LI", "CD274" and "B7-H1", and may be used interchangeably.
In the present invention, the term "antibody" is to be interpreted in its broadest sense and includes intact monoclonal antibodies, polyclonal antibodies, and multispecific antibodies (e.g., bispecific antibodies) formed from at least two intact antibodies, so long as they possess the desired biological activity. Herein, "antibody" and "immunoglobulin" may be used interchangeably.
In this context, the term "monoclonal antibody" means that the antibody is derived from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible natural mutations which may be present in minor amounts. A monoclonal antibody has a high specificity for one determinant (epitope) of an antigen, while a polyclonal antibody in contrast thereto comprises different antibodies directed against different determinants (epitopes). In addition to specificity, monoclonal antibodies also have the advantage that they can be synthesized without contamination by other antibodies. The modifier "monoclonal" used herein indicates that the antibody is characterized as being from a substantially homogeneous population of antibodies, and is not to be construed as requiring production by a particular method.
In some embodiments of the invention, monoclonal antibodies also specifically include chimeric antibodies, i.e., a portion of the heavy and/or light chain is identical or homologous to an antibody of one class, subclass, or subclass, and the remainder is identical or homologous to an antibody of another class, subclass, or subclass, so long as they possess the desired biological activity (see, e.g., U.S. Pat. No. 4,816,567; and Morrison et al, 1984, PNAS,81, 6851-6855. Chimeric antibodies useful in the invention include primatized antibodies comprising variable region antigen binding sequences from a non-human primate (e.g., an ancient monkey, orangutan, etc.) and human constant region sequences.
The term "antibody fragment" refers to a portion of an antibody, preferably the antigen binding or variable region. Examples of antibody fragments include Fab, fab ', F (ab') 2, fd, fv, dAb, and complementarity determining region fragments, antibodies (diabodies), linear antibodies, and single chain antibody molecules.
In the present invention, a "humanized" form of a non-human (e.g., murine) antibody refers to a chimeric antibody that contains minimal non-human immunoglobulin sequences. Most humanized antibodies are recipient immunoglobulin hypervariable region residues which are replaced with non-human (e.g., mouse, rat, rabbit or non-human primate) hypervariable region residues having the desired specificity, affinity, and function (donor antibody). In some embodiments, framework Region (FR) residues of the human immunoglobulin are also replaced with non-human residues. Furthermore, humanized antibodies may also comprise residues that are not present in the recipient antibody or the donor antibody. These modifications are made to further optimize the performance of the antibody. Humanized antibodies typically comprise at least one, and typically two, variable domains in which all or substantially all of the hypervariable loops (hypervariable loops) correspond to those of a non-human immunoglobulin and the FRs are fully or substantially fully sequences of a human immunoglobulin. The humanized antibody may also comprise at least a portion of an immunoglobulin constant region (Fc, typically a human immunoglobulin Fc). For details see, e.g., jones et al, 1986, nature, 321; riechmann et al, 1988, nature, 332; and Presta,1992, curr Op Struct Bwl 2.
Intact antibodies can be classified into different "classes" according to the amino acid sequence of the heavy chain constant region. The main five classes are IgA, igD, igE, igG and IgM, with several of the classes also being divided into different "subclasses" (isotypes), e.g. IgG1, igG2, igG3, igG4, igA1 and IgA2. The heavy chain constant regions of the different classes of antibodies are referred to as α, β, ε, γ, and μ, respectively. The different subunit structures and three-dimensional configurations of immunoglobulins are well known in the art.
Monoclonal antibodies used in the present invention can be produced by a number of methods. For example, monoclonal antibodies for use in the present invention can be obtained by hybridoma methods using a variety of species (including mouse, hamster, rat, and human cells) (see, e.g., kohler et al, 1975, nature, 256), or by recombinant DNA techniques (see, e.g., U.S. Pat. No. 4,816,567), or isolated from phage antibody libraries (see, e.g., clackson et al, 1991, nature, 352.
As used herein, "mammal" includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) or normal individuals suffering from a disease or disorder (e.g., a disease or disorder described herein). "non-human animals" in the context of the present invention include non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
Technical effects
The 1,2, 5-oxadiazole structure-containing compound useful as an IDO inhibitor of the present invention or its isomer, metabolite, stable isotope derivative, prodrug, pharmaceutically acceptable salt, hydrate, solvate, co-crystal or polymorph is used in combination with other prophylactic and/or therapeutic agents to achieve a good effect for preventing and/or treating diseases with abnormal cell proliferation. Experiments show that the treatment effect of the combined medicine group is better than that of the single medicine group, the synergistic and synergistic effect is achieved, and good tolerance is shown.
Detailed Description
The invention is further illustrated by the following description of the embodiments and the figures, which are not to be construed as limiting the invention. Various modifications and improvements may be made by those skilled in the art in light of the teachings of the invention without departing from the essential spirit and scope thereof.
Drawings
FIG. 1 is a schematic crystal structure of Compound 50-a.
Examples
Embodiments of the present invention will be described in detail below with reference to examples, but those skilled in the art will appreciate that the following examples are only illustrative of the present invention and should not be construed as limiting the scope of the present invention. The examples, in which specific conditions are not specified, were conducted under conventional conditions or conditions recommended by the manufacturer. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
Example 1 Synthesis of N- (2- ((4- (N- (3-bromo-4-fluorophenyl) -N '-hydroxycarbamimidoyl) -1,2, 5-oxadiazol-3-yl) amino) ethyl) -N' - (methylsulfonyl) morpholine-4-carboxamidine (Compound 50)
First step of
Figure BDA0002385910440000321
Compound 166c (100mg, 0.28mmol) was dissolved in DMF (5 mL), and triethylamine (0.18mL, 1.25mmol) and compound 134a (83mg, 0.42mmol) were added in this order and reacted at 70 ℃ for 2 hours. After disappearance of the starting material, concentration under reduced pressure gave compound 169a (124 mg, crude) which was used directly in the next step.
Second step of
Figure BDA0002385910440000322
Compound 169a (20mg, 0.04mmol), morpholine (7mg, 0.08mmol) and DIPEA (10mg, 0.08mmol) were dissolved in DMF (1 mL) and reacted at 100 ℃ for 4 hours. The reaction mixture was concentrated under reduced pressure and then separated and purified by high performance liquid chromatography to give compound 50 (16 mg, yield: 13.1%).
MS(ESI,m/z):549.4[M+H + ].
1 H NMR(400MHz,DMSO-d 6 )δ11.49(s,1H),8.88(s,1H),7.35(t,J=5.3Hz,1H),7.19(t,J=8.8Hz,1H),7.11(dt,J=7.2,3.6Hz,1H),6.79(ddd,J=8.8,4.1,2.8Hz,1H),6.44(t,J=5.7Hz,1H),3.57(m,4H),3.24(d,J=10.3Hz,3H),2.85(s,3H),1.23(s,4H).
EXAMPLE 2 determination of the stereostructure of Compound 50
5.1mg of Compound 50 was placed in a 3mL glass vial, 0.6mL of a mixed solvent of methanol/toluene (1, 3, v/v) was added, vortexed and suitably sonicated to facilitate solid dissolution, filtered using a PTFE frit (filter pore size 0.45 μm) and a 2.0mL syringe, the filtrate was transferred to a 4mL single crystal vial (44.6 mm. Times.14.65 mm), seeded with a small amount of the starting compound 50-a, sealed with a PE-Plug single crystal cap and perforated with a small hole, and placed in a fume hood for slow evaporation at room temperature. After slowly volatilizing for 4 days, a massive single crystal sample is observed and separated out from the system.
A single crystal with proper diffraction quality is cut and selected from the massive crystal sample, and the single crystal is wrapped with Paratone-N crystal protection oil (an oil-based crystal cryoprotectant) and adhered to a Loop ring. The single crystal samples were then mounted in random orientation on a crystal-holder table, and the crystals were immersed in ambient temperature (T = 296.15K). Single crystal diffraction data collection single crystal diffractometers of the Bruker D8VENTURE type (Mo target source,
Figure BDA0002385910440000332
) The analysis was performed at 296.15K temperature and the diffraction data was analyzed using the APEX3 software package. Each frame of diffraction image collected by the detector is subjected to reduction integration and lorentz polarization corrections (correction) by using a SAINT (Bruker, v8.38a, 2016) program, and 16774 diffraction points are collected, wherein each diffraction point is 5172 independent diffraction points. The diffraction data were absorption corrected using the SADABS-2016/2 (Bruker 2016/2) program (Multi-Scan method). Single crystal structure analysis the diffraction data were initially structure resolved (using the Intrinsic pharmacy method) using the ShelXT initial solution program using the OLEX2 software and the space group to which the crystals belong was determined. Followed by a structure refinement using the ShelXL (Version: 2017/1) program. And (3) determining the coordinates of all non-hydrogen atoms by using a number of rounds of difference Fourier synthesis, and then performing anisotropic refinement on all non-hydrogen atoms by using a full matrix least square method. All hydrogen atom coordinates were calculated by a theoretical hydrogenation (rating model).
Calculated XRPD spectra (Mo target X-rays) of single crystal structure data were calculated by the MerMory software based on atomic coordinates, space groups and unit cell parameters of the crystal structure.
The crystal structure schematic diagram is drawn by using Diamond software. A thermal vibration ellipsoid plot is plotted using ORTEP-III, as shown in FIG. 1.
The single crystal structure analysis confirmed that the stereochemistry of compound 50 was 50-a, and that two C = N double bonds { i.e., C (12) = N (8) and C (7) = N (2) double bonds } in the molecule were both Z-configuration.
Example 3N- (3-bromo-4-fluorophenyl) -4- ((2- ((1- (dimethylamino) -2-nitrovinyl) amino) ethyl) amino) -N' -hydroxy-1, 2, 5-oxadiazole-3-carboxamidine (Compound 35)
Figure BDA0002385910440000331
First step of
Compound 114a (210mg, 0.50mmol) was dissolved in methanol (5 mL), and 2.0M aqueous NaOH (0.5mL, 1.0 mmol) was added to the solution to react at room temperature for 0.5h. Work-up gave compound 166c (180 mg, crude) which was used directly in the next step.
Second step of
Compound 166c (100mg, 0.28mmol), 114b (67.3mg, 0.34mmol), N-diisopropylethylamine (0.11g, 0.84mmol), and N, N-dimethylformamide (5 mL) were added to a 50mL reaction flask, and the temperature was raised to 70 ℃ to react for 2 hours. The reaction mixture was cooled to room temperature, and dimethylamine aqueous solution (2 mL) was added thereto, and the temperature was raised to 55 ℃ to react overnight. The crude product obtained by the post-treatment was isolated and purified by high performance liquid chromatography to give the title compound 35 (33 mg, yield: 25.0%).
MS(ESI,m/z):473.1[M+H+]。
1H NMR(400MHz,DMSO-d 6 )δ11.36(s,1H),8.97(s,1H),8.46(s,1H),7.18(t,J=8.8Hz,1H),7.20–7.10(m,1H),6.85–6.71(m,1H),6.40–6.31(m,2H),3.51–3.43(m,4H),2.91(s,6H).
Experimental example 1 the composition of the present invention has drug effect on CT-26 mouse colon cancer transplantable tumor
In this experimental example, the efficacy of each test article was examined for changes in tumor volume and tumor weight in mice of a colon cancer transplantation tumor model in CT-26 mice after intraperitoneal injection (i.p.) of PD-1 antibody (Bioxcell, BP 0146) and intragastric administration (p.o.) of compound 35 or 50-a.
1.1 Experimental cell lines and Experimental animals
Experimental cell lines: mouse colon cancer CT-26 cells (Chinese academy of sciences type culture Collection cell Bank) were assayed at 37 ℃ and 5% CO 2 CulturingCultured in a chamber (medium: RPMI-1640 (Gibco, 31800) containing 10% fetal bovine serum (Biosera, FB-1058/500)). Passage was performed with conventional digestion treatment using pancreatin-EDTA (Hyclone, SH 30042.01). When the cells are in the exponential growth phase and the saturation is 80% -90%, collecting the cells and counting.
Experimental animals: BALB/c mice, 7 weeks old, female, 16-20 grams in weight, 48 in group, all purchased from shanghai slyke laboratory animals llc, all housed in a Special Pathogen Free (SPF) grade animal house.
1.2 tumor cell inoculation and grouping
CT-26 cells were resuspended in Dulbecco's Phosphate Buffer (DPBS) at a density of 3X 10 6 Individual cells/mL. 0.1mL of DPBS (containing 3X 10) 5 Individual CT26 cells) was subcutaneously inoculated on the right back of each mouse until the mean tumor volume reached 56mm 3 At time, groups were randomized into 6 according to tumor volume.
1.3 Experimental methods
The average volume of the subcutaneous transplanted tumor reaches 56mm 3 At that time, the tumor volume was randomly divided into 6 groups of 8 and administration was started for 21 days, and the specific administration schedule is shown in table 1. Tumor volume was measured 2 times a week after dosing, animals were observed daily for mortality, and tumor weight was measured after dosing was completed.
1.4 Experimental indices and statistical analysis
Tumor inhibiting therapeutic effect of test article from tumor volume inhibition rate TGI volume (%), relative tumor volume proliferation rate T/C volume (%), relative tumor weight proliferation rate T/C weight (%) or tumor weight inhibition ratio TGI weight And (6) evaluating.
Tumor volume was measured with a vernier caliper. The formula for tumor volume is: v =0.5 × a × b 2 And a and b represent the major and minor diameters of the tumor, respectively. Tumor volume inhibition ratio TGI volume (%) = [ (1- (average tumor volume at the end of administration in the administration group-average tumor volume at the start of administration in the administration group)/(average tumor volume at the end of administration in the vehicle control group-average tumor volume at the start of administration in the vehicle control group) ]]X100%. Relative tumor volume proliferation rateT/C volume (%)=T RTV /C RTV ×100%(T RTV : mean RTV of the groups administered; c RTV : vehicle control group mean RTV). Calculating Relative Tumor Volume (RTV) according to the measurement result of the tumor volume, wherein the calculation formula is RTV = V t /V 0 In which V is 0 Is a measure of the resulting tumor volume, V, upon divided administration t Tumor volume at a certain measurement, T RTV And C RTV The same day data was taken.
Tumor weights were measured after the end of the administration and the relative tumor weight proliferation rate T/C was calculated weight Percent, T/C weight (%)=TW treatment /TW Solvent ×100%,TW treatment And TW Solvent The tumor weights of the administered group and vehicle control group are shown, respectively. Tumor weight inhibition ratio TGI weight (%)=100%-T/C weight
The combined medication index calculates Q value according to the King's formula, Q is added when Q =0.85-1.15, and Q is added>1.15 is synergistic; q = E a+b /(E a +E b –E a ×E b ) In which E a+b Tumor inhibition Rate (TGI), E for the combination group a And E b Tumor inhibition rates (TGI) were given for the individual groups.
Statistical analysis was performed using SPSS software based on end of experiment versus tumor volume and tumor weight. Comparisons between two groups were analyzed using Student's t-test, comparisons between three or more groups were analyzed using one-way ANOVA, dunnett's method if the variance was uniform (F-values were not significantly different), and Games-Howell method if the variance was not uniform (F-values were significantly different). P <0.05 was considered a significant difference.
TABLE 1 test substance dosing regimens
Figure BDA0002385910440000351
Volume of administration: 10mL/kg.
Vehicle system a was 3% dmac (N, N-dimethylacetamide) +97% (10% hydroxypropyl- β cyclodextrin in water); vehicle system B was Dulbecco's Phosphate Buffered Saline (DPBS).
1.5 test results
After 21 days of administration, the weight average of animals in each administration group was increased and no animals died. The results of the test for tumor weight, tumor volume and tumor inhibitory effect in mice are shown in tables 2 to 5.
Figure BDA0002385910440000361
TABLE 3 Effect of test Compound 35 on tumor weight in CT-26 Homoplastic mice
Figure BDA0002385910440000371
Note: a. tumor weight (g) = mean ± Standard Error (SEM), n =8.
b. Relative tumor weight gain rate T/C weight (%)=TW treatment /TW Solvent ×100%。
c. Tumor weight inhibition rate TGI weight (%)=100%-T/C weight
Q = Ea + b/(Ea + Eb-Ea × Eb), wherein Ea + b is tumor weight inhibition ratio (TGI) of combined drug weight ) Ea and Eb are tumor weight inhibition rates (TGI) of the single drug groups weight )。
P values were obtained by analyzing tumor weight using one-way ANOVA and vehicle treatment groups. According to the Laviny statistics (Leven static) that F values are significantly different (P = 0.040), the analysis is performed by using the Games-Howell method.
After 21 days of administration, when Compound 35 (100 mg/kg) was administered in combination with the PD-1 antibody (5 mg/kg), the tumor volume growth inhibition ratio TGI volume 76.74% (P =0.049, vs. vehicle group), relative tumor weight gain rate T/C weight 23.25% (P =0.005, vs. vehicle group), tumor weight inhibition rate TGI weight It was 76.75%. According to the tumor volume and the tumor weight data, the combined medication index Q of the tumor volume and the tumor weight is respectively calculated to be 1.58 and 1.95 by a King's formula. The above results indicate that Compound 35 is synergistic with PD-1 antibody (5 mg/kg) at 100 mg/kg.
Figure BDA0002385910440000381
TABLE 5 Effect of test Compound 50-a on tumor weight in CT26 Homoplastic mice
Figure BDA0002385910440000391
Note: a. tumor weight (g) = mean ± SEM, n =8.
b. Relative tumor weight growth inhibition rate T/C weight (%)=TW treatment /TW Solvent ×100%。
c. Tumor weight inhibition ratio TGI weight (%)=100%-T/Cweight。
Q = Ea + b/(Ea + Eb-Ea × Eb), wherein Ea + b is tumor weight inhibition rate (TGI) of the combined drug weight ) Ea and Eb are the tumor weight inhibition rates (TGI) of the respective drug groups weight )。
P values were obtained by analyzing tumor weight using one-way ANOVA and vehicle treatment groups. According to the Laviny statistics (Leven static) that F values are significantly different (P = 0.040), the analysis is performed by using the Games-Howell method.
After 21 days of administration, compound 50-a (50 mg/kg) when administered in combination with PD-1 antibody (5 mg/kg) showed a tumor volume growth inhibition rate TGI volume 75.05% (P =0.037, vs. vehicle group), relative tumor weight gain rate T/C weight 29.27% (P =0.009, vs. vehicle group), tumor weight inhibition rate TGI weight It was 70.73%. Simultaneously, according to the tumor volume and the tumor weight data, the combined medication index Q of the tumor volume and the tumor weight is respectively calculated to be 1 through a King's formula22 and 1.40. The above results indicate that Compound 50-a is synergistic with the PD-1 antibody (5 mg/kg) at 50 mg/kg.

Claims (8)

1. Use of a compound containing a1, 2, 5-oxadiazole structure, or a pharmaceutically acceptable salt thereof, in combination with one or more other prophylactic and/or therapeutic agents in the manufacture of a medicament for the prevention and/or treatment of colon cancer; the compound containing a1, 2, 5-oxadiazole structure is selected from the group consisting of:
Figure FDA0003946025270000011
the other prophylactic and/or therapeutic drug is a PD-1 or PD-L1 antibody.
2. The use according to claim 1, said other prophylactic and/or therapeutic drug being a PD-1 antibody.
3. Use of compound 50-a or a pharmaceutically acceptable salt thereof and a PD-1 antibody in the manufacture of a medicament for the prevention and/or treatment of colon cancer;
Figure FDA0003946025270000012
4. use of compound 35 or a pharmaceutically acceptable salt thereof and a PD-1 antibody in the manufacture of a medicament for the prevention and/or treatment of colon cancer;
Figure FDA0003946025270000013
5. a pharmaceutical composition comprising a compound containing the 1,2, 5-oxadiazole structure or a pharmaceutically acceptable salt thereof as the active ingredient, together with one or more other prophylactic and/or therapeutic agents, and a pharmaceutically acceptable carrier or excipient; the compound containing a1, 2, 5-oxadiazole structure is selected from:
Figure FDA0003946025270000014
the other prophylactic and/or therapeutic drug is selected from a PD-1 antibody or a PD-L1 antibody.
6. The composition according to claim 5, wherein the other prophylactic and/or therapeutic agent is a PD-1 antibody.
7. A method for preparing a medicament suitable for combined use, comprising formulating a prophylactically and/or therapeutically effective amount of a compound containing a1, 2, 5-oxadiazole structure or a pharmaceutically acceptable salt thereof and one or more other prophylactic and/or therapeutic agents into a medicament for the prophylaxis and/or treatment of colon cancer, which is suitable for combined use or administration; the compound containing a1, 2, 5-oxadiazole structure is selected from:
Figure FDA0003946025270000021
the other prophylactic and/or therapeutic drug is selected from a PD-1 antibody or a PD-L1 antibody.
8. The method according to claim 7, wherein said other prophylactic and/or therapeutic agent is a PD-1 antibody.
CN202010098154.4A 2019-02-28 2020-02-18 Pharmaceutical compositions, methods and uses thereof for the treatment and/or prevention of disease Active CN111617078B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019101495610 2019-02-28
CN201910149561 2019-02-28

Publications (2)

Publication Number Publication Date
CN111617078A CN111617078A (en) 2020-09-04
CN111617078B true CN111617078B (en) 2023-02-03

Family

ID=72267021

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010098154.4A Active CN111617078B (en) 2019-02-28 2020-02-18 Pharmaceutical compositions, methods and uses thereof for the treatment and/or prevention of disease

Country Status (1)

Country Link
CN (1) CN111617078B (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112194723B (en) * 2020-09-25 2021-09-21 广州百吉生物制药有限公司 Application of immune cells in treating cancer
CN112315954A (en) * 2020-11-24 2021-02-05 烟台大学 Paclitaxel and IDO1 small molecule inhibitor compound pharmaceutical composition and application thereof
CN115227812A (en) * 2021-04-22 2022-10-25 上海君实生物医药科技股份有限公司 Use of anti-PD-1 antibody in combination with first-line chemotherapy for treating advanced NSCLC
CN114409610B (en) * 2022-03-29 2022-06-10 山东大学 Oxadiazole derivatives, their preparation and use
CN114767678A (en) * 2022-03-31 2022-07-22 武汉大学中南医院 Use of Linrodostatt in preparation of medicine for treating bladder cancer
CN115089719A (en) * 2022-06-15 2022-09-23 苏州大学 Synergistic ovarian cancer immunotherapy pharmaceutical composition and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104042611A (en) * 2008-07-08 2014-09-17 因塞特公司 1,2,5-oxadiazoles As Inhibitors Of Indoleamine 2,3-dioxygenase
WO2019011170A1 (en) * 2017-07-14 2019-01-17 四川科伦博泰生物医药股份有限公司 Oxadiazole derivative, preparation method therefor and medical application thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2744880C1 (en) * 2014-02-04 2021-03-16 Инсайт Корпорейшн Combination of pd-1 antagonist and ido1 inhibitor for treating ancer
WO2018028491A1 (en) * 2016-08-09 2018-02-15 苏州国匡医药科技有限公司 Indoleamine2,3-dioxygenase inhibitors and uses thereof in pharmacy
CN108341777A (en) * 2017-01-23 2018-07-31 四川科伦博泰生物医药股份有限公司 Compound of isobioquin group and its application

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104042611A (en) * 2008-07-08 2014-09-17 因塞特公司 1,2,5-oxadiazoles As Inhibitors Of Indoleamine 2,3-dioxygenase
WO2019011170A1 (en) * 2017-07-14 2019-01-17 四川科伦博泰生物医药股份有限公司 Oxadiazole derivative, preparation method therefor and medical application thereof

Also Published As

Publication number Publication date
CN111617078A (en) 2020-09-04

Similar Documents

Publication Publication Date Title
CN111617078B (en) Pharmaceutical compositions, methods and uses thereof for the treatment and/or prevention of disease
CN109890387B (en) Use of glutamate modulators and immunotherapy for the treatment of cancer
US20210137955A1 (en) Combination therapy for cancer treatment
CN112566661B (en) Pharmaceutical combination of quinoline derivatives with antibodies
BR112019021880A2 (en) COMBINATION THERAPY WITH ANTI-AXL-DRUG ANTIBODY CONJUGATE
TWI685341B (en) Use of combination of apatinib and c-met inhibitor in the preparation of medicament for treating tumor
AU2020228056A1 (en) Compounds with ferroptosis inducing activity and methods of their use
TW201938149A (en) Combination of a selective histone deacetylase 3 (HDAC3) inhibitor and an immunotherapy agent for the treatment of cancer
WO2022133731A1 (en) Pharmaceutical combinations comprising a kras g12c inhibitor and uses of a kras g12c inhibitor and for the treatment of cancers
CN113811298B (en) Quinoline derivatives for the combined treatment of small cell lung cancer
JP2022189827A (en) Administration of hypoxia activated prodrugs in combination with immune modulatory agents for treating cancer
AU2015240775B2 (en) Sigma-2 receptor ligand drug conjugates as antitumor compounds, methods of synthesis and uses thereof
TW202216698A (en) Solid forms of a cdk4 inhibitor
US20210046082A1 (en) Crystalline forms of quinoline analogs and salts thereof, compositions, and their methods for use
EP3421039B1 (en) Drug for cancer therapy characterized by administering combination between axl inhibitor and immune checkpoint inhibitor
CN110831604B (en) Pharmaceutical composition, method and use for tumor treatment or prevention
CA2944255C (en) New derivatives of cephalosporin for treating cancer
JPWO2019039525A1 (en) Cancer therapeutic agent containing an Axl inhibitor as an active ingredient
CA3121441C (en) Ezh1/2 dual inhibitor-containing pharmaceutical composition to be used as a combination drug
EP3973963A1 (en) Quinoline derivatives for treatment of head and neck cancer
CA3150514A1 (en) Treatment of cancer using a combination comprising multi-tyrosine kinase inhibitor and immune checkpoint inhibitor
TW202045175A (en) Administration of sumo-activating enzyme inhibitor and checkpoint inhibitors
EP4134098A1 (en) Method of cancer therapy
WO2024015637A1 (en) Glutathione peroxidase 4 (gpx4) inhibitors for the treatment of cancer

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant